US9056865B2 - Pyridine-2-derivatives as smoothened receptor modulators - Google Patents

Pyridine-2-derivatives as smoothened receptor modulators Download PDF

Info

Publication number
US9056865B2
US9056865B2 US13/880,298 US201113880298A US9056865B2 US 9056865 B2 US9056865 B2 US 9056865B2 US 201113880298 A US201113880298 A US 201113880298A US 9056865 B2 US9056865 B2 US 9056865B2
Authority
US
United States
Prior art keywords
chloro
bipyridin
methyl
piperidin
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US13/880,298
Other languages
English (en)
Other versions
US20130210800A1 (en
Inventor
Sajiv Krishnan Nair
Simon Paul Planken
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Priority to US13/880,298 priority Critical patent/US9056865B2/en
Publication of US20130210800A1 publication Critical patent/US20130210800A1/en
Application granted granted Critical
Publication of US9056865B2 publication Critical patent/US9056865B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems

Definitions

  • Embodiments disclosed herein relate to novel pyridine derivatives that are useful in therapy, in particular for treating diseases or conditions mediated by Smo, including the treatment of abnormal cell growth, such as cancer, in mammals. Additional embodiments relate to methods of using such compounds in the treatment of abnormal cell growth in mammals, especially humans, and to pharmaceutical compositions containing such compounds.
  • Hedgehog (Hh) proteins are secreted morphogens that are involved in many biological processes during embryonic development. Postnatally, Hh has important roles in tissue homeostasis and aberrant Hh signaling is associated with developmental disorders and several types of cancer. At the cell surface, the Hh signal is thought to be relayed by the 12 transmembrane domain protein Patched (Ptc) (Hooper and Scott, Cell 59: 75 1-65 (1989); Nakano et al., Nature 341: 508-13 (1989)) and the G-protein-coupled-like receptor Smoothened (Smo) (Alcedo et al., Cell 86: 221-232 (1996); van den Heuvel and Tngham, Nature 382: 547-551 (1996)).
  • Ptc transmembrane domain protein Patched
  • Smo G-protein-coupled-like receptor Smoothened
  • the signaling cascade initiated by Smo results in activation of Gli transcription factors that translocate into the nucleus where they control transcription of target genes.
  • Gli has been shown to influence transcription of Hh pathway inhibitors such as Ptc and Hip I in a negative feedback loop indicating that tight control of the Hh pathway activity is required for proper cellular differentiation and organ formation.
  • Uncontrolled activation of Hh signaling pathway is associated with malignancies in particular those of the brain, skin and muscle as well as angiogenesis.
  • An explanation for this is that the Hh pathway has been shown to regulate cell proliferation in adults by activation of genes involved in cell cycle progression such as cyclin D which is involved in G1-S transition.
  • Sonic Hedgehog (SHh), an ortholog of Hh, blocks cell-cycle arrest mediated by p21, an inhibitor of cyclin dependent kinases.
  • Hh signaling is further implicated in cancer by inducing components in the EGFR pathway (EGF, Her2) involved in proliferation as well as components in the PDGF (PDGFa) and VEGF pathways involved in angiogenesis.
  • EGF epidermal growth factor
  • PDGFa vascular endothelial growth factor
  • VEGFa vascular endothelial growth factor pathway
  • Loss of function mutations in the Ptc gene have been identified in patients with the basal cell nevus syndrome (BCNS), a hereditary disease characterized by multiple basal cell carcinomas (BCCs).
  • BCNS basal cell nevus syndrome
  • BCCs basal cell carcinomas
  • Dysfunctional Ptc gene mutations have also been associated with a large percentage of sporadic basal cell carcinoma tumors (Chidambaram et al., Cancer Research 56: 4599-601 (1996); Gailani et al., Nature Genet. 14: 78-81 (1996); Hahn et al., Cell 85: 841-51 (1996); Johnson et al., Science 272: 1668-71 (1996); Unden et al., Cancer Res. 56: 4562-5; Wicking et al., Am. J. Hum. Genet. 60: 21-6 (1997)). Loss of Ptc function is thought to cause an uncontrolled Smo signaling in basal cell carcinoma.
  • A is selected from N and C—R 13 ;
  • X and Y are independently selected from N and C—R 12 , provided that at least one of X and Y is N;
  • R 1 , R 2 , R 3 , R 4 , R 6 , R 12 and R 13 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O) 2 R 16
  • R 5 is selected from halo, C 1 -C 10 alkyl, and —CF 3 ,
  • each R 7 , R 8 , R 9 , and R 10 is independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O) 2 R 16 , —(CR 14 R 15
  • R 11 is selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR A R B , —(CR 14 R 15 ) n NR A OR B , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n NR A C(O)OR B , —(CR 14 R 15 ) n OR A , —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n C(O)OR A ,
  • R A and R B are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n NR 16 OR 17 , —(CR 14 R 15 ) n NR 16 C(O)R 17 , —(CR 14 R 15 ) n NR 16 C(O)OR 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)
  • each R 14 , R 15 , R 16 and R 17 is independently selected from hydrogen, —(CR 24 R 25 ) n halo, —(CR 24 R 25 ) n CF 3 , —(CR 24 R 25 ) n (C 1 -C 10 alkyl), —(CR 24 R 25 ) n (C 2 -C 6 alkenyl), —(CR 24 R 25 ) n (C 2 -C 6 alkynyl), —(CR 24 R 25 ) n OR 18 , —(CR 24 R 25 ) n NR 18 R 19 , —(CR 24 R 28 ) n CN, —(CR 24 R 25 ) n S(O) 2 R 18 , —(CR 24 R 25 ) n S(O) 2 NR 18 R 19 , —(CR 24 R 25 ) n (C 3 -C 10 cycloalkyl), —(CR 24 R 25 ) n (3-12 membered heterocycly
  • each R 18 , R 19 , R 24 and R 25 is independently selected from hydrogen, —(CH 2 ) n (C 1 -C 10 alkyl), —(CH 2 ) n (C 3 -C 10 cycloalkyl), —(CH 2 ) n (3-12 membered heterocyclyl), —(CH 2 ) n (C 6 -C 10 aryl), and —(CH 2 ) n (5-12 membered heteroaryl);
  • e 1 or 2;
  • f 1 or 2;
  • each n is independently selected from 0, 1, 2, 3, 4, 5, and 6;
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), and —(CR 14 R 15 ) n OR 16 .
  • Additional embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • each R 7 , R 8 , R 9 , and R 10 is independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), and —(CR 14 R 15 ) n OR 16 .
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein each R 7 , R 8 , R 9 , and R 10 is independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • More embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein e is 2 and f is 2.
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 5 is halo.
  • Additional embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is N; Y is C—R 12 ; and R 12 is selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • More embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is N and Y is N.
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n S(O) 2 R A , —(CR 14 R 15 ) n S(O) 2 NR A R B , and —(CR 14 R 15 ) n NR A S(O) 2 R B .
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein A is N.
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —C(O)R A and —S(O) 2 R A .
  • Additional embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 5 is F or Cl; R 11 is C(O)R A ; and R A is —(CR 14 R 15 ) n S(O) 2 R 16 .
  • the compound of formula (I) is:
  • the compound of formula (I) is:
  • Additional embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein A is C—R 13 .
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —NR A C(O)R B and —NR A S(O) 2 R B .
  • More embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 11 is NR A S(O) 2 R B .
  • Some embodiments relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R A is hydrogen and R 5 is F or Cl.
  • the compound of formula (I) is:
  • X and Y are independently selected from N and C—R 12 , provided that at least one of X and Y is N;
  • Z is selected from NR 11b and CR 13 NR 14 R 11b ;
  • R 1 , R 2 , R 3 , R 4 , R 6b , R 6 , R 12 and R 13 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O
  • each R 20 , R 21 , R 22 and R 23 is independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O) 2 R 16 , —(CR 14 R 15 )
  • R 11b is selected from C(O)R A and S(O) 2 R A ;
  • R A is selected from —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n NR 16 OR 17 , —(CR 14 R 15 ) n NR 16 C(O)R 17 , —(CR 14 R 15 ) n NR 16 C(O)OR 17 , —(CR 14 R 15 ) n NR 16 S(O) 2 R 17 , —(CR 14 R 15 ) n (C 3 -C 10 cycloalkyl), —(CR 14 R 15 ) n (3-12 membered heterocyclyl), —(CR 14 R 15 ) n (C 6
  • each R 14 , R 15 , R 16 and R 17 is independently selected from hydrogen, —(CR 24 R 25 ) n halo, —(CR 24 R 25 ) n CF 3 , —(CR 24 R 25 ) n C 1 -C 10 alkyl, —(CR 24 R 25 ) n C 2 -C 6 alkenyl, —(CR 24 R 25 ) n C 2 -C 6 alkynyl, —(CR 24 R 25 ) n OR 18 , —(CR 24 R 25 ) n NR 18 R 19 , —(CR 24 R 25 ) n CN, (CR 24 R 25 ) n S(O) 2 R 18 , —(CR 24 R 25 ) n S(O) 2 NR 18 R 19 , —(CR 24 R 25 ) n C 3 -C 10 cycloalkyl, —(CR 24 R 25 ) n 3-12 membered heterocyclyl, —(CR
  • each R 18 , R 19 , R 24 and R 25 is independently selected from hydrogen, —(CH 2 ) n (C 1 -C 10 alkyl), —(CH 2 ) n (C 3 -C 10 cycloalkyl), —(CH 2 ) n (3-12 membered heterocyclyl), —(CH 2 ) n (C 6 -C 10 aryl), and —(CH 2 ) n (5-12 membered heteroaryl);
  • f 2;
  • each n is independently selected from 0, 1, 2, 3, 4, 5, and 6;
  • each m is independently selected from 1, 2, 3, 4, 5, and 6;
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), and —(CR 14 R 15 ) n OR 16 .
  • Additional embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • More embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein each R 20 , R 21 , R 22 , and R 23 is independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), and —(CR 14 R 15 ) n OR 16 .
  • each R 20 , R 21 , R 22 , and R 23 is independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • Yet additional embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 5b is selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), and —(CR 14 R 15 ) n OR 16 .
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 5b is selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 5b is selected from hydrogen, bromine, chlorine, and fluorine.
  • Additional embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is N; Y is C—R 12 ; and R 12 is selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • Still more embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is N and Y is N.
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein Z is CR 13 NR 14 R 11b .
  • Additional embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein Z is NR 11b .
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 5b is selected from hydrogen, chlorine, and fluorine; R 11b is S(O) 2 R A ; R A ; R A is selected from —(CR 14 R 15 ) n C 1 -C 10 alkyl), —(CR 14 R 15 ) m OR 16 , and —(CR 14 R 15 ) m C(O)R 16 ; and R 16 is selected from hydrogen, —CF 3 , and C 1 -C 10 alkyl.
  • Some embodiments relate to a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 5b is selected from hydrogen, chlorine, and fluorine; R 11b is C(O)R A ; and R A is —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) m OR 16 , and —(CR 14 R 15 ) m C(O)R 16 , and —(CR 14 R 15 ) m S(O) 2 R 16 .
  • the compound of formula (II) is:
  • R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O) 2 R 16 , —(CR 14
  • R 5 is selected from halo, C 1 -C 10 alkyl, and —CF 3 ,
  • R 11 is selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR A R B , —(CR 14 R 15 ) n NR A OR B , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n NR A C(O)OR B , —(CR 14 R 15 ) n OR A , —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n C(O)OR A ,
  • R A and R B are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n NR 16 OR 17 , —(CR 14 R 15 ) n NR 16 C(O)R 17 , —(CR 14 R 15 ) n NR 16 C(O)OR 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)
  • each R 14 , R 15 , R 16 and R 17 is independently selected from hydrogen, —(CR 24 R 25 ) n halo, —(CR 24 R 25 ) n CF 3 , —(CR 24 R 25 ) n (C 1 -C 10 alkyl), —(CR 24 R 25 ) n (C 2 -C 6 alkenyl), —(CR 24 R 25 ) n (C 2 -C 6 alkynyl), —(CR 24 R 25 ) n OR 18 , —(CR 24 R 25 ) n NR 18 R 19 , —(CR 24 R 25 ) n CN, —(CR 24 R 25 ) n S(O) 2 R 18 , —(CR 24 R 25 ) n S(O) 2 NR 18 R 19 , —(CR 24 R 25 ) n (C 3 -C 10 cycloalkyl), —(CR 24 R 25 ) n (3-12 membered heterocycly
  • each R 18 , R 19 , R 24 and R 25 is independently selected from hydrogen, —(CH 2 ) n (C 1 -C 10 alkyl), —(CH 2 ) n (C 3 -C 10 cycloalkyl), —(CH 2 ) n (3-12 membered heterocyclyl), —(CH 2 ) n (C 6 -C 10 aryl), and —(CH 2 ) n (5-12 membered heteroaryl); and
  • each n is independently selected from 0, 1, 2, 3, 4, 5, and 6;
  • Some embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • Additional embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 5 is halo.
  • More embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n S(O) 2 R A , —(CR 14 R 15 ) n S(O) 2 NR A R B , and —(CR 14 R 15 ) n NR A S(O) 2 R B .
  • Some embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —C(O)R A and —S(O) 2 R A .
  • Some embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 11 is C(O)R A .
  • Additional embodiments relate to a compound of formula (III), or a pharmaceutically acceptable salt thereof, wherein R 5 is F or Cl; R 11 is C(O)R A ; and R A is —(CR 14 R 15 ) n S(O) 2 R 16 .
  • R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)OR 16 , —(CR 14 R 15 ) n S(O)R 16 , —(CR 14 R 15 ) n S(O) 2 R 16 , —(CR 14
  • R 5 is selected from halo, C 1 -C 10 alkyl, and —CF 3 ,
  • R 11 is selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR A R B , —(CR 14 R 15 ) n NR A OR B , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n NR A C(O)OR B , —(CR 14 R 15 ) n OR A , —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n C(O)OR A ,
  • R A and R B are independently selected from hydrogen, —(CR 14 R 15 ) n halo, —(CR 14 R 15 ) n CN, —(CR 14 R 15 ) n CF 3 , —(CR 14 R 15 ) n (C 1 -C 10 alkyl), —(CR 14 R 15 ) n (C 2 -C 6 alkenyl), —(CR 14 R 15 ) n (C 2 -C 6 alkynyl), —(CR 14 R 15 ) n NR 16 R 17 , —(CR 14 R 15 ) n NR 16 OR 17 , —(CR 14 R 15 ) n NR 16 C(O)R 17 , —(CR 14 R 15 ) n NR 16 C(O)OR 17 , —(CR 14 R 15 ) n OR 16 , —(CR 14 R 15 ) n C(O)R 16 , —(CR 14 R 15 ) n C(O)
  • each R 14 , R 15 , R 16 and R 17 is independently selected from hydrogen, —(CR 24 R 25 ) n halo, —(CR 24 R 25 ) n CF 3 , —(CR 24 R 25 ) n (C 1 -C 10 alkyl), —(CR 24 R 25 ) n (C 2 -C 6 alkenyl), —(CR 24 R 25 ) n (C 2 -C 6 alkynyl), —(CR 24 R 25 ) n OR 18 , —(CR 24 R 25 ) n NR 18 R 19 , —(CR 24 R 25 ) n CN, —(CR 24 R 25 ) n S(O) 2 R 18 , —(CR 24 R 25 ) n S(O) 2 NR 18 R 19 , —(CR 24 R 25 ) n (C 3 -C 10 cycloalkyl), —(CR 24 R 25 ) n (3-12 membered heterocycly
  • each R 18 , R 19 , R 24 and R 25 is independently selected from hydrogen, —(CH 2 ) n (C 1 -C 10 alkyl), —(CH 2 ) n (C 3 -C 10 cycloalkyl), —(CH 2 ) n (3-12 membered heterocyclyl), —(CH 2 ) n (C 6 -C 10 aryl), and —(CH 2 ) n (5-12 membered heteroaryl); and
  • each n is independently selected from 0, 1, 2, 3, 4, 5, and 6;
  • Some embodiments relate to a compound of formula (IV), or a pharmaceutically acceptable salt thereof, wherein R 1 , R 2 , R 3 , R 4 , and R 6 are independently selected from hydrogen, -halo, —CN, —CF 3 , and —(C 1 -C 10 alkyl).
  • Additional embodiments relate to a compound of formula (IV), or a pharmaceutically acceptable salt thereof, wherein R 5 is halo.
  • More embodiments relate to a compound of formula (IV), or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from —(CR 14 R 15 ) n C(O)R A , —(CR 14 R 15 ) n NR A C(O)R B , —(CR 14 R 15 ) n S(O) 2 R A , —(CR 14 R 15 ) n S(O) 2 NR A R B , and —(CR 14 R 15 ) n NR A S(O) 2 R B .
  • Some embodiments relate to a compound of formula (IV), or a pharmaceutically acceptable salt thereof, wherein R 11 is —S(O) 2 R A .
  • Additional embodiments relate to a compound of formula (IV), or a pharmaceutically acceptable salt thereof, wherein R 5 is Cl and R 11 is —S(O) 2 R A .
  • Additional embodiments relate to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), (II), (Ill), or (IV), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or diluent.
  • said cancer is selected from the group consisting of basal cell cancer, medulloblastoma cancer, liver cancer, rhabdomyosarcoma, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the
  • the compound is selected from:
  • C 1 -C 10 alkyl refers to saturated monovalent hydrocarbon radicals containing from one to ten carbon atoms, having straight or branched moieties.
  • C 3 -C 10 cycloalkyl refers to a mono, fused or bridged bicyclic or tricyclic carbocyclic rings containing from three to ten carbon atoms, (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl and bicyclo[5.2.0]nonanyl, norbornyl, adamantanyl, etc.); said rings may optionally contain 1 or 2 double bonds.
  • cycloalkyl also includes spiro cycloalkyl groups, including multi-ring systems joined by a single atom.
  • carrier refers to an aliphatic ring system having three to twelve members.
  • carrier refers to rings that are optionally substituted.
  • carrier also include aliphatic rings that are fused to one or more aromatic or non-aromatic rings, such as in a decahydronaphthyl or tetrahydronaphthyl, where the radical or point-of attachment is on the aliphatic ring.
  • C 2 -C 6 alkenyl used alone or as part of a larger moiety shall include both straight and branched chains containing two to six carbon atoms having at least one carbon-carbon double bond.
  • C 2 -C 6 alkenyl used alone or as part of a larger moiety shall include both straight and branched chains containing two to ten carbon atoms having at least one carbon-carbon triple bond.
  • halo is used herein interchangeably with the term “halogen”, which denotes F, Cl, Br, or I. Preferred halo groups are F, Cl, and Br.
  • heteroatom refers to nitrogen, oxygen, or sulfur and includes any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen.
  • nitrogen includes a substitutable nitrogen of a heterocyclic ring.
  • the nitrogen in a saturated or partially unsaturated ring having 0 to 3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NOR (as in N-substituted pyrrolidinyl).
  • C 6 -C 10 aryl refers to a group derived from an aromatic hydrocarbon containing from 6 to 10 carbon atoms. Examples of such groups include, but are not limited to, phenyl and naphthyl.
  • benzyl refers to a —CH 2 C 6 H 5 group.
  • C 6 -C 10 aryl also includes fused polycyclic aromatic ring systems in which an aromatic ring is fused to one or more rings.
  • C 6 -C 10 aryl examples include 1-naphthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl.
  • C 6 -C 10 aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as in an indanyl, phenanthridinyl, or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic ring.
  • C 6 -C 10 aryl also refers to rings that are optionally substituted.
  • heteroaryl used alone or as part of a larger moiety as in “heteroaralkyl” or “heteroarylalkoxy”, refers to an aromatic heterocyclic group having a total of from 5 to 12 atoms in its ring, and containing from 2 to 9 carbon atoms and from one to four heteroatoms each independently selected from O, S and N, with the proviso that the ring of said group does not contain two adjacent O atoms or two adjacent S atoms.
  • the heterocyclic groups include benzo-fused ring systems.
  • aromatic heterocyclic groups include, but are not limited to, pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quina
  • heteroaryl is a group in which a heteroatomic ring is fused to one or more aromatic or nonaromatic rings where the radical or point of attachment is on the heteroaromatic ring.
  • examples include tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[3,4-d]pyrimidinyl.
  • heterocyclyl also known as heterocycle, or heteroalicyclic refers to a non-aromatic, monocyclic, bicyclic, tricyclic or spirocyclic ring group having a total of 3 to 12 ring atoms, in which 1 to 4 ring atoms are heteroatoms selected from N, O, and S, and wherein the S atom may be optionally oxidized with one or two oxygen atoms, the remaining ring atoms being C, with the proviso that such ring systems may not contain two adjacent O atoms or two adjacent S atoms.
  • the heterocyclic ring may also be substituted by an oxo ( ⁇ O) group at any available C atom.
  • the rings may also have one or more double bonds. Furthermore, such groups may be bonded to the remainder of the compounds of embodiments disclosed herein through either a carbon atom or a heteroatom, if possible.
  • the heterocylic ring may also be benzo-fused, where the point of attachment is on the heterocyclic ring. Examples of suitable saturated heterocyclyl groups include, but are not limited to:
  • Suitable partially unsaturated heterocyclyl groups include, but are not limited to:
  • heterocyclyl or “heterocycle”, as previously noted, also includes spirocyclic moieties containing at least one heteroatom in one or more of the spirocyclic rings (also known as “heterospirocyclic” or “heterospirocyclic ring”). Such heterospirocyclic moieties may be optionally substituted at any ring position, including substitution on the heteratom(s) within the spirocyclic ring(s). Examples of spirocyclic moieties include, but are not limited to:
  • treating means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of treating as “treating” is defined immediately above.
  • an “effective” amount refers to an amount of a substance, agent, compound, or composition that is of sufficient quantity to result in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction—either as a single dose or according to a multiple dose regimen, alone or in combination with other agents or substances.
  • One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.
  • the subject may be a human or non-human mammal (e.g., rabbit, rat, mouse, monkey or other lower-order primate).
  • Embodiments disclosed herein include isotopically-labeled compounds, which are identical to those recited in formula I or formula II, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the embodiments disclosed herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • Isotopically-labeled compounds of embodiments disclosed herein can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically-labeled reagent for a non-isotopically-labeled reagent.
  • Some embodiments also relate to the pharmaceutically acceptable acid addition salts of the compounds described herein.
  • the acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned base compounds are those which form nontoxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as, but not limited to, the chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1′ methylene bis(2 hydroxy 3 naphthoate)]salts.
  • nontoxic acid addition salts i.e
  • Additional embodiments relate to base addition salts of the compounds described herein.
  • the chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of those compounds of the compounds described herein that are acidic in nature are those that form non-toxic base salts with such compounds.
  • Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., potassium and sodium) and alkaline earth metal cations (e.g., calcium and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines.
  • phrases “pharmaceutically acceptable salt(s)”, as used herein, unless otherwise indicated, includes salts of acidic or basic groups which may be present in the compounds of described herein.
  • the compounds described herein that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds described herein are those that form non-toxic acid addition salts, e.g., salts containing pharmacologically acceptable anions, such as the hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate)
  • the compounds of the embodiments described herein include all stereoisomers (e.g., cis and trans isomers) and all optical isomers of compounds described herein (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers. While all stereoisomers are encompassed within the scope of our claims, one skilled in the art will recognize that particular stereoisomers may be preferred.
  • the compounds described herein can exist in several tautomeric forms, including the enol and imine form, and the keto and enamine form and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the present embodiments. Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present embodiments includes all tautomers of the present compounds.
  • the present embodiments also include atropisomers of the compounds described herein.
  • Atropisomers refer to compounds that can be separated into rotationally restricted isomers.
  • compositions described herein also relate to the pharmaceutically acceptable salts of the compounds described herein.
  • Pharmaceutically acceptable salts of the compounds described herein include the acid addition and base salts thereof.
  • Suitable acid addition salts are formed from acids which form non-toxic salts.
  • suitable acid addition salts include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyrogluta
  • Suitable base salts are formed from bases which form non-toxic salts.
  • suitable base salts include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
  • Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • the compounds described herein may also exist in unsolvated and solvated forms. Accordingly, some embodiments relate to the hydrates and solvates of the compounds described herein.
  • solvate is used herein to describe a molecular complex comprising a compound described herein and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds described herein containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. A single compound may exhibit more than one type of isomerism.
  • Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
  • the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where a compound described herein contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • a suitable optically active compound for example, an alcohol, or, in the case where a compound described herein contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Further embodiments relate to methods of treating abnormal cell growth in a mammal. Additional embodiments relate to a method of treating abnormal cell growth in a mammal comprising administering to said mammal an amount of a compound described herein that is effective in treating abnormal cell growth.
  • the abnormal cell growth is cancer.
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (
  • Additional embodiments relate to methods of treating cancer solid tumors in a mammal. Some embodiments relate to the treatment of cancer solid tumor in a mammal comprising administering to said mammal an amount of a compound described herein that is effective in treating said cancer solid tumor.
  • the cancer solid tumor is breast, lung, colon, brain, prostate, stomach, pancreatic, ovarian, skin (melanoma), endocrine, uterine, testicular, or bladder.
  • compositions for treating abnormal cell growth in a mammal comprising an amount of a compound described herein that is effective in treating abnormal cell growth, and a pharmaceutically acceptable carrier.
  • Additional embodiments relate to a method of treating abnormal cell growth in a mammal, including a human, comprising administering to said mammal an amount of a compound described herein, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, that is effective in treating abnormal cell growth.
  • the abnormal cell growth is cancer, including, but not limited to, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms
  • the method comprises comprising administering to a mammal an amount of a compound described herein that is effective in treating said cancer solid tumor.
  • the solid tumor is breast, lung, colon, brain, prostate, stomach, pancreatic, ovarian, skin (melanoma), endocrine, uterine, testicular, and bladder cancer.
  • said abnormal cell growth is a benign proliferative disease, including, but not limited to, psoriasis, benign prostatic hypertrophy or restinosis.
  • Some embodiments relate to a method of treating abnormal cell growth in a mammal which comprises administering to said mammal an amount of a compound described herein, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, that is effective in treating abnormal cell growth in combination with an anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, antibodies, cytotoxics, anti-hormones, and anti-androgens.
  • an anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, antibodies, cytotoxics, anti-hormones, and anti-androgens.
  • Additional embodiments relate to a pharmaceutical composition for treating abnormal cell growth in a mammal, including a human, comprising an amount of a compound described herein, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, that is effective in treating abnormal cell growth, and a pharmaceutically acceptable carrier.
  • said abnormal cell growth is cancer, including, but not limited to, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms
  • compositions for treating abnormal cell growth contemplate a pharmaceutical composition for treating abnormal cell growth wherein the composition includes a compound described herein, or a pharmaceutically acceptable salt, solvate, or hydrate thereof, that is effective in treating abnormal cell growth, and another anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, antibodies, cytotoxics, anti-hormones, and anti-androgens.
  • another anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, antibodies, cytotoxics, anti-hormones, and anti-androgens.
  • Yet more embodiments relate to a method of treating a disorder associated with angiogenesis in a mammal, including a human, comprising administering to said mammal an amount of a compound described herein, as defined above, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, that is effective in treating said disorder in combination with one or more anti-tumor agents listed above.
  • Such disorders include cancerous tumors such as melanoma; ocular disorders such as age-related macular degeneration, presumed ocular histoplasmosis syndrome, and retinal neovascularization from proliferative diabetic retinopathy; rheumatoid arthritis; bone loss disorders such as osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, hypercalcemia from tumors metastatic to bone, and osteoporosis induced by glucocorticoid treatment; coronary restenosis; and certain microbial infections including those associated with microbial pathogens selected from adenovirus, hantaviruses, Borrelia burgdorferi, Yersinia spp., Bordetella pertussis , and group A Streptococcus.
  • Some embodiments relate to a method of (and to a pharmaceutical composition for) treating abnormal cell growth in a mammal which comprise an amount of a compound described herein, or a pharmaceutically acceptable salt, solvate, or hydrate thereof, in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors inhibitor (e.g., inhibiting the means by which regulatory molecules that govern the fundamental processes of cell growth, differentiation, and survival communicated within the cell), and antiproliferative agents, which amounts are together effective in treating said abnormal cell growth.
  • signal transduction inhibitors inhibitor e.g., inhibiting the means by which regulatory molecules that govern the fundamental processes of cell growth, differentiation, and survival communicated within the cell
  • antiproliferative agents which amounts are together effective in treating said abnormal cell growth.
  • Anti-angiogenesis agents such as MMP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors, can be used in conjunction with a compound described herein in the methods and pharmaceutical compositions described herein.
  • MMP-2 matrix-metalloprotienase 2
  • MMP-9 matrix-metalloprotienase 9 inhibitors
  • COX-II cyclooxygenase II
  • Examples of useful COX-II inhibitors include CELEBREXTM (celecoxib), Bextra (valdecoxib), paracoxib, Vioxx (rofecoxib), and Arcoxia (etoricoxib).
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published Oct. 24, 1996), WO 96/27583 (published Mar.
  • MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or MMP-9 relative to the other matrix-metalloproteinases (i.e. MMP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13).
  • MMP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13 matrix-metalloproteinases
  • MMP inhibitors useful in combination with the compounds described herein are AG-3340, RO 32-3555, RS 13-0830, and the following compounds:
  • VEGF inhibitors for example, SU-11248, SU-5416 and SU-6668 (Sugen Inc. of South San Francisco, Calif., USA), can also be combined with a compound described herein.
  • VEGF inhibitors are described in, for example in WO 99/24440 (published May 20, 1999), PCT International Application PCT/IB99/00797 (filed May 3, 1999), in WO 95/21613 (published Aug. 17, 1995), WO 99/61422 (published Dec. 2, 1999), U.S. Pat. No. 5,834,504 (issued Nov. 10, 1998), WO 98/50356 (published Nov. 12, 1998), U.S. Pat. No. 5,883,113 (issued Mar. 16, 1999), U.S. Pat. No.
  • VEGF inhibitors include IM862 (Cytran Inc. of Kirkland, Wash., USA); Avastin, an anti-VEGF monoclonal antibody of Genentech, Inc. of South San Francisco, Calif.; and angiozyme, a synthetic ribozyme from Ribozyme (Boulder, Colo.) and Chiron (Emeryville, Calif.).
  • ErbB2 receptor inhibitors such as GW-282974 (Glaxo Wellcome plc), and the monoclonal antibodies AR-209 (Aronex Pharmaceuticals Inc. of The Woodlands, Tex., USA) and 2B-1 (Chiron), may be administered in combination with a compound described herein.
  • erbB2 inhibitors include Herceptin, 2C4, and pertuzumab.
  • Such erbB2 inhibitors include those described in WO 98/02434 (published Jan. 22, 1998), WO 99/35146 (published Jul. 15, 1999), WO 99/35132 (published Jul. 15, 1999), WO 98/02437 (published Jan. 22, 1998), WO 97/13760 (published Apr.
  • ErbB2 receptor inhibitors useful in the embodiments described herein are also described in U.S. Provisional Application No. 60/117,341, filed Jan. 27, 1999, and in U.S. Provisional Application No. 60/117,346, filed Jan. 27, 1999, both of which are herein incorporated by reference in their entirety.
  • Other erbb2 receptor inhibitors include TAK-165 (Takeda) and GW-572016 (Glaxo-Wellcome).
  • World Patent Application WO 92/20642 (published Nov. 26, 1992), refers to certain bis-mono and bicyclic aryl and heteroaryl compounds as tyrosine kinase inhibitors that are useful in inhibiting abnormal cell proliferation.
  • World Patent Applications WO96/16960 (published Jun. 6, 1996), WO 96/09294 (published Mar. 6, 1996), WO 97/30034 (published Aug. 21, 1997), WO 98/02434 (published Jan. 22, 1998), WO 98/02437 (published Jan.
  • antiproliferative agents that may be used with the compounds described herein include inhibitors of the enzyme farnesyl protein transferase and inhibitors of the receptor tyrosine kinase PDGFr, including the compounds disclosed and claimed in the following U.S. patent application Ser. Nos. 09/221,946 (filed Dec. 28, 1998); 09/454,058 (filed Dec. 2, 1999); 09/501,163 (filed Feb. 9, 2000); 09/539,930 (filed Mar. 31, 2000); 09/202,796 (filed May 22, 1997); 09/384,339 (filed Aug. 26, 1999); and 09/383,755 (filed Aug. 26, 1999); and the compounds disclosed and claimed in the following U.S.
  • a compound described herein may also be used with other agents useful in treating abnormal cell growth or cancer, including, but not limited to, agents capable of enhancing antitumor immune responses, such as CTLA4 (cytotoxic lymphocyte antigen 4) antibodies, and other agents capable of blocking CTLA4; and anti-proliferative agents such as other farnesyl protein transferase inhibitors, for example the farnesyl protein transferase inhibitors described in the references cited in the “Background” section, supra.
  • CTLA4 cytotoxic lymphocyte antigen 4
  • anti-proliferative agents such as other farnesyl protein transferase inhibitors, for example the farnesyl protein transferase inhibitors described in the references cited in the “Background” section, supra.
  • Specific CTLA4 antibodies that can be used in the present embodiments include those described in U.S. Provisional Application 60/113,647 (filed Dec. 23, 1998) which is herein incorporated by reference in its entirety.
  • a compound described herein may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, for example, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis-platin, oxaliplatin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, capecitabine, cytosine arabinoside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • mitotic inhibitors for example vinblastine
  • alkylating agents for example cis-platin, oxaliplatin, carboplatin and cyclophosphamide
  • anti-metabolites for example 5-fluorouracil, capecitabine, cytosine arabinoside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • the compounds described herein may be used alone or in combination with one or more of a variety of anti-cancer agents or supportive care agents.
  • the compounds described herein may be used with cytotoxic agents, e.g., one or more selected from the group consisting of a camptothecin, irinotecan HCl (Camptosar), edotecarin, SU-11248, epirubicin (Ellence), docetaxel (Taxotere), paclitaxel, rituximab (Rituxan) bevacizumab (Avastin), imatinib mesylate (Gleevac), Erbitux, gefitinib (Iressa), and combinations thereof.
  • cytotoxic agents e.g., one or more selected from the group consisting of a camptothecin, irinotecan HCl (Camptosar), edotecarin, SU-11248, epirubicin (Ellence), docetaxe
  • Some embodiments also contemplate the use of the compounds described herein together with hormonal therapy, e.g., exemestane (Aromasin), Lupron, anastrozole (Arimidex), tamoxifen citrate (Nolvadex), Trelstar, and combinations thereof.
  • hormonal therapy e.g., exemestane (Aromasin), Lupron, anastrozole (Arimidex), tamoxifen citrate (Nolvadex), Trelstar, and combinations thereof.
  • some embodiments provide a compound described herein alone or in combination with one or more supportive care products, e.g., a product selected from the group consisting of Filgrastim (Neupogen), ondansetron (Zofran), Fragmin, Procrit, Aloxi, Emend, or combinations thereof.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • the compounds described herein may be used with antitumor agents, alkylating agents, antimetabolites, antibiotics, plant-derived antitumor agents, camptothecin derivatives, tyrosine kinase inhibitors, antibodies, interferons, and/or biological response modifiers.
  • antitumor agents alkylating agents, antimetabolites, antibiotics, plant-derived antitumor agents, camptothecin derivatives, tyrosine kinase inhibitors, antibodies, interferons, and/or biological response modifiers.
  • secondary agents that may be used with the compounds described herein.
  • Alkylating agents include, but are not limited to, nitrogen mustard N-oxide, cyclophosphamide, ifosfamide, melphalan, busulfan, mitobronitol, carboquone, thiotepa, ranimustine, nimustine, temozolomide, AMD-473, altretamine, AP-5280, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, ifosfamide, KW-2170, mafosfamide, and mitolactol; platinum-coordinated alkylating compounds include but are not limited to, cisplatin, carboplatin, eptaplatin, lobaplatin, nedaplatin, oxaliplatin or satrplatin.
  • Antimetabolites include but are not limited to, methotrexate, 6-mercaptopurine riboside, mercaptopurine, 5-fluorouracil (5-FU) alone or in combination with leucovorin, tegafur, UFT, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, S-1, gemcitabine, fludarabin, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethynylcytidine, cytosine arabinoside, hydroxyurea, TS-1, melphalan, nelarabine, nolatrexed, ocfosfate, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincri
  • Antibiotics include but are not limited to: aclarubicin, actinomycin D, amrubicin, annamycin, bleomycin, daunorubicin, doxorubicin, elsamitrucin, epirubicin, galarubicin, idarubicin, mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, valrubicin or zinostatin.
  • Hormonal therapy agents e.g., exemestane (Aromasin), Lupron, anastrozole (Arimidex), doxercalciferol, fadrozole, formestane, anti-estrogens such as tamoxifen citrate (Nolvadex) and fulvestrant, Trelstar, toremifene, raloxifene, lasofoxifene, letrozole (Femara), or anti-androgens such as bicalutamide, flutamide, mifepristone, nilutamide, Casodex® (4′-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3′-(trifluoromethyl)propionanilide) and combinations thereof.
  • Plant derived anti-tumor substances include for example those selected from mitotic inhibitors, for example vinblastine, docetaxel (Taxotere) and paclitaxel.
  • Cytotoxic topoisomerase inhibiting agents include one or more agents selected from the group consisting of aclarubicn, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan HCl (Camptosar), edotecarin, epirubicin (Ellence), etoposide, exatecan, gimatecan, lurtotecan, mitoxantrone, pirarubicin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide, and topotecan, and combinations thereof.
  • Interferons include interferon alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon gamma-1a or interferon gamma-n1.
  • agents include PF3512676, filgrastim, lentinan, sizofilan, TheraCys, ubenimex, WF-10, aldesleukin, alemtuzumab, BAM-002, dacarbazine, daclizumab, denileukin, gemtuzumab ozogamicin, ibritumomab, imiquimod, lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, OncoVAX-CL, sargramostim, tasonermin, tecleukin, thymalasin, tositumomab, Virulizin, Z-100, epratuzumab, mitumomab, oregovomab, pemtumomab, Provenge.
  • Biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity.
  • agents include krestin, lentinan, sizofiran, picibanil, or ubenimex.
  • anticancer agents include alitretinoin, ampligen, atrasentan bexarotene, bortezomib. Bosentan, calcitriol, exisulind, finasteride, fotemustine, ibandronic acid, miltefosine, mitoxantrone, I-asparaginase, procarbazine, dacarbazine, hydroxycarbamide, pegaspargase, pentostatin, tazarotne, TLK-286, Velcade, Tarceva, or tretinoin.
  • anti-angiogenic compounds include acitretin, fenretinide, thalidomide, zoledronic acid, angiostatin, aplidine, cilengtide, combretastatin A-4, endostatin, halofuginone, rebimastat, removab, Revlimid, squalamine, ukrain and Vitaxin.
  • Platinum-coordinated compounds include but are not limited to, cisplatin, carboplatin, nedaplatin, or oxaliplatin.
  • Camptothecin derivatives include but are not limited to camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, irinotecan, SN-38, edotecarin, and topotecan.
  • Tyrosine kinase inhibitors include, for example, Iressa and SU5416.
  • Antibodies include, for example, Herceptin, Erbitux, Avastin, and Rituximab.
  • Interferons include, for example, interferon alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon gamma-1a and interferon gamma-n1.
  • Biological response modifiers include agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity.
  • agents include, for example, krestin, lentinan, sizofiran, picibanil, and ubenimex.
  • antitumor agents include, for example, mitoxantrone, 1-asparaginase, procarbazine, dacarbazine, hydroxycarbamide, pentostatin, and tretinoin.
  • the compounds described herein are potent inhibitors of Smo, and thus are all adapted to therapeutic use as antiproliferative agents (e.g., anticancer), antitumor (e.g., effective against solid tumors), antiangiogenesis (e.g., stop or prevent proliferation of blood vessels) in mammals, particularly in humans.
  • antiproliferative agents e.g., anticancer
  • antitumor e.g., effective against solid tumors
  • antiangiogenesis e.g., stop or prevent proliferation of blood vessels
  • the compounds described herein are useful in the prevention and treatment of a variety of human hyperproliferative disorders such as malignant and benign tumors of the liver, kidney, bladder, breast, gastric, ovarian, colorectal, prostate, pancreatic, lung, vulval, thyroid, hepatic carcinomas, sarcomas, glioblastomas, head and neck, and other hyperplastic conditions such as benign hyperplasia of the skin (e.g., psoriasis) and benign hyperplasia of the prostate (e.g., BPH). It is, in addition, expected that a compound described herein may possess activity against a range of leukemias and lymphoid malignancies.
  • cancer is lung cancer, bone cancer, pancreatic cancer, gastric, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, gynecological, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, squamous cell, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, ne
  • cancer is selected a solid tumor, such as, but not limited to, breast, lung, colon, brain (e.g., glioblastoma), prostate, stomach, pancreatic, ovarian, skin (melanoma), endocrine, uterine, testicular, and bladder.
  • brain e.g., glioblastoma
  • prostate e.g., prostate, stomach, pancreatic, ovarian
  • skin melanoma
  • endocrine e.g., uterine, testicular, and bladder.
  • the methods described herein include the use of small molecules which inhibit Smo, in the regulation of repair and/or functional performance of a wide range of cells, tissues and organs, including normal cells, tissues, and organs, as well as those having the phenotype of ptc loss-of-function, hedgehog gain-of-function, or smoothened gain-of-function.
  • the subject method has therapeutic and cosmetic applications ranging from regulation of neural tissues, bone and cartilage formation and repair, regulation of spermatogenesis, regulation of smooth muscle, regulation of lung, liver and other organs arising from the primative gut, regulation of hematopoietic function, regulation of skin and hair growth, etc.
  • the subject methods can be performed on cells that are provided in culture (in vitro), or on cells in a whole animal (in vivo). See, for example, PCT publications WO 95/18856 and WO 96/17924.
  • Some embodiments also relate to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or formula II, or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • compositions which comprises mixing a compound of formula I or formula II, or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the daily dosage of the compound of formula I or formula II, or pharmaceutically acceptable salt thereof may be in the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg.
  • the present embodiments also encompass sustained release compositions.
  • Administration of the compounds described herein can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), topical, and rectal administration.
  • the active compound may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, for example, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis-platin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, cytosine arabinoside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • mitotic inhibitors for example vinblastine
  • alkylating agents for example cis-platin, carboplatin and cyclophosphamide
  • anti-metabolites for example 5-fluorouracil, cytosine arabinoside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • the pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound described herein as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compounds in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents.
  • the pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, binders, excipients and the like.
  • excipients such as citric acid
  • disintegrants such as starch, alginic acid and certain complex silicates
  • binding agents such as sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes.
  • Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules.
  • Preferred materials include lactose or milk sugar and high molecular weight polyethylene glycols.
  • the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • the compounds described herein may be prepared by processes known in the chemical arts, particularly in light of the description contained herein. Certain processes for the manufacture of the compounds described herein are provided as further features of the embodiments and are illustrated in the reaction schemes provided below and in the experimental section.
  • Et 2 O diethyl ether
  • DMF N,N-dimethylformamide
  • THF tetrahydrofuran
  • DCM diichloromethane
  • DMA dimethyl acetal
  • DBU (1,8-diazabicyclo[5.4.0]undec-7-ene)
  • HATU (2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate methanaminium
  • LDA lithium diisopropylamide
  • DMSO dimethylsulfoxide
  • DIPEA N,N-diisopropylethylamine
  • mCPBA metala-chloroperoxybenzoic acid
  • TFA trifluoroacetic acid
  • dppf (1,1′-bis(diphenylphosphino)ferrocene
  • heteroaryl boronic acid 1 can be treated with an aryl- or heteroaryl halide or aryl- or hetereoaryl trifluoromethylsulfonate 2 in a transition metal mediated Suzuki coupling to provide chloropyridine 3.
  • heteroaryl halide or trifluoromethane sulfonate 5 can be reacted with an aryl- or heteroaryl boronic acid or boronic ester, an aryl- or heteroaryl stannane, or an aryl- or heteroaryl zincate 4 in a transition metal catalyzed Suzuki, Stille or Negishi coupling to provide 3.
  • R 5 can be Cl, F, Br, Alkyl, or OR;
  • R 33 can be B(OR) 2 , SnR 3 , or ZnX;
  • LG can be Br, I or OTf;
  • R 32 can be Br, I or OTf;
  • R 30 and R 31 together with the nitrogen to which they are attached, may combine to form a heterocycle as depicted in formula (I) or formula (II).
  • Step 1 2′,5′-dichloro-3,5-dimethyl-2,4′-bipyridine
  • Step 2 Tert-butyl 4-(5′-chloro-3,5-dimethyl-2,4′-bipyridin-2′-yl)piperazine-1-carboxylate
  • Step 4 1-(4-(5′-chloro-3,5-dimethyl-2,4′-bipyridin-2′-yl)piperazin-1-yl)-3-(methylsulfonyl)propan-1-one
  • Step 6 5-chloro-4-(3-methylpyridin-2-yl)-2-(methylsulfonyl)pyrimidine and 5-chloro-4-(3-methylpyridin-2-yl)-2-(methylsulfinyl)pyrimidine
  • Step 7 5-chloro-4-(3-methylpyridin-2-yl)-2-(4-(methylsulfonylmethyl)piperidin-1-yl)pyrimidine
  • NCS (370 mg, 2.83 mmol) was added to a solution of N-(1-(4-(pyridin-2-yl)pyrimidin-2-yl)piperidin-4-yl)methanesulfonamide (180 mg, 2.36 mmol) in AcOH (10 mL) at 60° C. and the reaction solution was stirred for 2 h. LC-MS indicated the reaction was almost complete. The reaction solution was concentrated and purified by chromatography, eluted with CH 2 Cl 2 :MeOH (6:1) to give crude product, which was further purified by Prep-HPLC to give title compound (251 mg, 29%) as a yellow solid.
  • acylating agents such as activated carboxylic acids or acyl chlorides, carbamoyl chlorides, isocyanates and sulfonyl chlorides can provide amides, carbamates, ureas, and sulfonamides.
  • Step 5 3-chloro-6-[4-(vinylsulfonyl)piperazin-1-yl]-2,2′-bipyridine
  • Step 6 3-chloro-6- ⁇ 4-[(2-methoxyethyl)sulfonyl]piperazin-1-yl ⁇ -2,2′-bipyridine
  • Step 7 2- ⁇ [4-(3-chloro-2,2′-bipyridin-6-yl)piperazin-1-yl]sulfonyl ⁇ ethanol
  • Scheme A-5 An alternative approach to amino pyridines similar to those highlighted in Scheme A-4 is illustrated in Scheme A-5.
  • Treatment of 3,5-dimethylpyridine with m-CPBA gives the corresponding pyridine N-oxide.
  • Deprotonation of the N-oxide followed by iodination and subsequent reduction of the N-oxide moiety with phoshporous tribromide provides 2-iodo-3,5-dimethylpyridine.
  • Cross-coupling of this iodide with 2-bromo-6-chloropyridine under Negishi coupling conditions gives 6′-chloro-3,5-dimethyl-2,2′-bipyridine.
  • a palladium-catalyzed C—H halogenation of this 2,2′-bipyridine using palladium acetate and N-chlorosuccinimide provides the 3′,6′-dichloro-3,5-dimethyl-2,2′-bipyridine.
  • Treatment of this dichloride with suitably protected or unprotected amines in the presence of a base such as cesium fluoride in a suitable solvent (such as DMSO) can yield final products.
  • acylating agents such as activated carboxylic acids or acyl chlorides and sulfonyl chlorides can provide amides and sulfonamides.
  • Step 6 tert-butyl [1-(3-chloro-3′,5′-dimethyl-2,2′-bipyridin-6-yl)piperidin-4-yl]carbamate
  • Step 7 1-(3-chloro-3′,5′-dimethyl-2,2′-bipyridin-6-yl)piperidin-4-amine (hydrochloride salt)
  • Step 8 N-[1-(3-chloro-3′,5′-dimethyl-2,2′-bipyridin-6-yl)piperidin-4-yl]-2-hydroxyacetamide
  • Step 1 Tert-butyl 1-(5′-chloro-3′-fluoro-3,5-dimethyl-2,4′-bipyridin-2′-yl)piperidin-4-ylcarbamate
  • Step 3 N-(1-(5′-chloro-3′-fluoro-3,5-dimethyl-2,4′-bipyridin-2′-yl)piperidin-4-yl)-3-(methylsulfonyl)propanamide
  • Membranes were prepared from a stable cell line created in HEK293FlpIn-TetR cells (Invitrogen) using Flp recombinase-mediated insertion of the pSecTag-FRT/V5-His vector containing a cDNA encoding amino acids 181-787 of human Smo fused to the murine Igk leader sequence to produce a cell surface expressed Smo 181-781 protein.
  • Hygromycin-resistant clones were obtained and stained for LacZ expression (no expression indicates a correct knock-in of fusion cDNA). LacZ-negative cells were analyzed for binding tritiated Smo antagonists.
  • the HEK293 cells expressing Smo 181-781 were grown to 90% confluence in nine to fifteen 245 mm ⁇ 245 mm ⁇ 22 mm dishes, washed with Dulbecco's PBS (15 mL per dish) and harvested via scraping in 10 mL of DPBS. The cells were collected and centrifuged at 1500 rpm (400 ⁇ g) for 10 min at 4° C. The cell pellets were re-suspended in 40 mL of cold DPBS and washed by centrifugation at 2300 rpm (950 ⁇ g max) for 10 minutes at 4° C.
  • This homogenate was diluted up to 50 mL using Membrane Preparation Buffer and centrifuged at 35,000 rpm in a Beckman Ti45 rotor (140,000 ⁇ g) for 35 minutes at 4° C. followed by aspiration of the supernatant and re-suspension of the pellet in 5 mL of Assay Buffer (50 mM Tris-HCl pH 7.5, 100 mM NaCl, 25 mM MgCl 2 , 1 mM EDTA, and 0.1% protease free bovine serum albumin). The re-suspended pellet was then homogenized in a glass tissue grinder. The re-suspended membranes were aliquoted (0.5 mL aliquots), snap frozen and stored at ⁇ 70° C. Total protein in the membrane preparation was determined using the Pierce BCA protein assay (Pierce Chemical).
  • Assay Buffer 100 ⁇ L of Assay Buffer was added to all the wells of a 96 well GF/B filter plate (Millipore MultiScreen-HTS-FB cat# MSFBN6B50) for 10 minutes to pre-wet the filter prior to evacuation of the buffer (8 inches Hg for 8 seconds).
  • 20 ⁇ L of Assay Buffer 10 ⁇ L diluted test agent, 20 ⁇ L of a tritiated Smo antagonist (15 nM stock solution), and 50 ⁇ L of membrane preparation (40 ⁇ g total protein per well).
  • the plates are sealed and mixed at room temperature for 5 min, incubated at room temperature for 2 hours, then washed 5 times with 100 ⁇ L/each of wash buffer and vacuum dried for 8 seconds at 8 inches Hg.
  • the plate is then dried for one hour in a 60° C. oven prior to the addition of 45 ⁇ L of Microscint 20 (Packard, #6013621) to each well and incubation at RT for 30 minutes to 1 hour.
  • the plate is counted in a TopCount scintillation counter (Perkin Elmer).
  • X is the logarithm of the inhibitor concentration.
  • Y is the response; Y starts at Bottom and goes to Top with a sigmoid shape.
  • the Gli-Luc/MEF cells obtained from Gli-Luc transgenic mice contain a luciferase reporter gene under the control of the Gli response element. Luciferase activity stimulated with Sonic hedgehog ligand was inhibited by Smo inhibitors, and IC 50 was subsequently calculated.
  • Gli-Luc/MEF cells were grown in Knockout DMEM media (Invitrogen 10829-18) supplemented with 10% Heat inactive Fetal Bovine Serum (FBS, Hyclone), 2 mM L-glutamine (Invitrogen 25030-80), and 0.55 mM ⁇ -mercaptoethanol) until 90° A) confluence.
  • FBS Heat inactive Fetal Bovine Serum
  • 2 mM L-glutamine Invitrogen 25030-80
  • 0.55 mM ⁇ -mercaptoethanol 0.55 mM ⁇ -mercaptoethanol
  • Bright-Glo luciferase reagent was made up and 25 ⁇ L were added to each well of the 384-well plate containing media. Plates were kept at room temperature for 5 minutes, and then read on an Envision Luminescence plate reader (Perkin-Elmer). IC 50 of the inhibition was calculated by using GraphPad Prism.
  • the total plasma concentrations were 2240 ⁇ 296 ng/mL, 2220 ⁇ 1780 ng/mL, and 287 ⁇ 75 ng/mL at 1 hour, 4 hours and 7 hours, respectively.
  • the total plasma concentrations were multiplied by 0.067, a previously determined unbound fraction of rat plasma protein binding.
  • the resulting unbound plasma concentrations were 150 ⁇ 20 ng/mL (1 hour), 149 ⁇ 119 ng/mL (4 hours), and 19.2 ⁇ 5.0 ng/mL (7 hours).
  • the respective CSF concentrations were 64.4 ⁇ 12.0 ng/mL, 72.1 ⁇ 71.9 ng/mL, and 3.9 ⁇ 2.3 ng/mL at 1 hour, 4 hours, and 7 hours post dose.
  • A-116 is not a P-glycoprotein substrate
  • the CSF concentration was used as a surrogate for unbound concentrations in the brain.
  • a ratio of the CSF concentration to the unbound plasma concentration was calculated. At 1 hour, 4 hours, and 7 hours, the ratios were 0.43 ⁇ 0.03, 0.44 ⁇ 0.1, and 0.19 ⁇ 0.06, respectively.
  • the study demonstrated that a significant amount of A-116, approximately 40% of unbound A-116 in the plasma, was able to cross the blood brain barrier in rats (within 4 hours post dose).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
US13/880,298 2010-10-20 2011-10-19 Pyridine-2-derivatives as smoothened receptor modulators Expired - Fee Related US9056865B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/880,298 US9056865B2 (en) 2010-10-20 2011-10-19 Pyridine-2-derivatives as smoothened receptor modulators

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US40510110P 2010-10-20 2010-10-20
PCT/IB2011/054675 WO2012052948A1 (fr) 2010-10-20 2011-10-19 Dérivés de pyridine-2 en tant que modulateurs des récepteurs smoothened
US13/880,298 US9056865B2 (en) 2010-10-20 2011-10-19 Pyridine-2-derivatives as smoothened receptor modulators

Publications (2)

Publication Number Publication Date
US20130210800A1 US20130210800A1 (en) 2013-08-15
US9056865B2 true US9056865B2 (en) 2015-06-16

Family

ID=44936320

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/880,298 Expired - Fee Related US9056865B2 (en) 2010-10-20 2011-10-19 Pyridine-2-derivatives as smoothened receptor modulators

Country Status (6)

Country Link
US (1) US9056865B2 (fr)
EP (1) EP2630134B9 (fr)
JP (1) JP5802756B2 (fr)
CA (1) CA2813162C (fr)
ES (1) ES2543151T3 (fr)
WO (1) WO2012052948A1 (fr)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8628554B2 (en) 2010-06-13 2014-01-14 Virender K. Sharma Intragastric device for treating obesity
US9526648B2 (en) 2010-06-13 2016-12-27 Synerz Medical, Inc. Intragastric device for treating obesity
US10010439B2 (en) 2010-06-13 2018-07-03 Synerz Medical, Inc. Intragastric device for treating obesity
US10420665B2 (en) 2010-06-13 2019-09-24 W. L. Gore & Associates, Inc. Intragastric device for treating obesity
EP2723732B1 (fr) 2011-06-22 2017-01-18 Purdue Pharma LP Antagonistes de trpv1 comprenant un substituant dihydroxy et leurs utilisations
WO2014113191A1 (fr) * 2013-01-15 2014-07-24 Xiaohu Zhang Inhibiteurs de la voie signalisation hedgehog et leurs applications thérapeutiques
CN103588771B (zh) * 2013-01-15 2016-01-27 苏州云轩医药科技有限公司 具有刺猬通路拮抗剂活性的嘧啶类抗肿瘤化合物
CN103333117B (zh) * 2013-05-18 2015-11-25 嘉兴中科化学有限公司 一种4[(4-氯-2-嘧啶基)氨基]苯腈的制备方法
US9884865B2 (en) 2013-08-26 2018-02-06 Purdue Pharma L.P. Azaspiro[4.5] decane derivatives and use thereof
JO3517B1 (ar) 2014-01-17 2020-07-05 Novartis Ag ان-ازاسبيرو الكان حلقي كبديل مركبات اريل-ان مغايرة وتركيبات لتثبيط نشاط shp2
CN105899491B (zh) 2014-01-17 2019-04-02 诺华股份有限公司 用于抑制shp2活性的1-哒嗪-/三嗪-3-基-哌(-嗪)/啶/吡咯烷衍生物及其组合物
US9815813B2 (en) 2014-01-17 2017-11-14 Novartis Ag 1-(triazin-3-yl/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions therefor for inhibiting the activity of SHP2
WO2016203404A1 (fr) 2015-06-19 2016-12-22 Novartis Ag Composés et compositions pour inhiber l'activité de shp2
JP6878316B2 (ja) 2015-06-19 2021-05-26 ノバルティス アーゲー Shp2の活性を阻害するための化合物および組成物
WO2016203406A1 (fr) 2015-06-19 2016-12-22 Novartis Ag Composés et compositions pour inhiber l'activité de shp2
US10779980B2 (en) 2016-04-27 2020-09-22 Synerz Medical, Inc. Intragastric device for treating obesity
US10934285B2 (en) 2016-06-14 2021-03-02 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
BR112019000213B1 (pt) * 2016-07-05 2022-10-25 Bayer Cropscience Aktiengesellschaft Derivados de piridina halogenados, seu processo de preparação e seus intermediários
IL305530A (en) * 2021-03-18 2023-10-01 Sironax Ltd Inhibitors of receptor-binding protein 1, their preparations and their uses

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009154739A2 (fr) 2008-06-17 2009-12-23 Duke University Modulateurs du récepteur smoothened
US20100029615A1 (en) 2006-12-15 2010-02-04 Michael John Munchhof Benzimidazole derivatives
US8148401B2 (en) 2007-06-29 2012-04-03 Pfizer Inc. Benzimidazole derivatives
US20120157471A1 (en) 2009-09-01 2012-06-21 Pfizer Inc. Benzimidazole derivatives

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8607683D0 (en) 1986-03-27 1986-04-30 Ici Plc Anti-tumor agents
GB8827305D0 (en) 1988-11-23 1988-12-29 British Bio Technology Compounds
AU658646B2 (en) 1991-05-10 1995-04-27 Rhone-Poulenc Rorer International (Holdings) Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
NZ243082A (en) 1991-06-28 1995-02-24 Ici Plc 4-anilino-quinazoline derivatives; pharmaceutical compositions, preparatory processes, and use thereof
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
AU661533B2 (en) 1992-01-20 1995-07-27 Astrazeneca Ab Quinazoline derivatives
CA2129663C (fr) 1992-02-06 2005-07-05 James S. Huston Proteine fixatrice biosynthetique pour marqueur du cancer
US6177401B1 (en) 1992-11-13 2001-01-23 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften Use of organic compounds for the inhibition of Flk-1 mediated vasculogenesis and angiogenesis
GB9323290D0 (en) 1992-12-10 1994-01-05 Zeneca Ltd Quinazoline derivatives
US5455258A (en) 1993-01-06 1995-10-03 Ciba-Geigy Corporation Arylsulfonamido-substituted hydroxamic acids
GB9314893D0 (en) 1993-07-19 1993-09-01 Zeneca Ltd Quinazoline derivatives
GB9314884D0 (en) 1993-07-19 1993-09-01 Zeneca Ltd Tricyclic derivatives
WO1995018856A1 (fr) 1993-12-30 1995-07-13 President And Fellows Of Harvard College Proteines de type herisson induisant une structure embryonnaire chez les vertebres
IL112248A0 (en) 1994-01-25 1995-03-30 Warner Lambert Co Tricyclic heteroaromatic compounds and pharmaceutical compositions containing them
GB9510757D0 (en) 1994-09-19 1995-07-19 Wellcome Found Therapeuticaly active compounds
GB9424233D0 (en) 1994-11-30 1995-01-18 Zeneca Ltd Quinazoline derivatives
US6281332B1 (en) 1994-12-02 2001-08-28 The Johns Hopkins University School Of Medicine Hedgehog-derived polypeptides
US5863949A (en) 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
DK0821671T3 (da) 1995-04-20 2001-04-23 Pfizer Arylsulfonylhydroxamsyrederivater som MMP- og TNF-inhibitorer
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
GB9520822D0 (en) 1995-10-11 1995-12-13 Wellcome Found Therapeutically active compounds
GB9624482D0 (en) 1995-12-18 1997-01-15 Zeneca Phaema S A Chemical compounds
DK0780386T3 (da) 1995-12-20 2003-02-03 Hoffmann La Roche Matrixmetalloproteaseinhibitorer
GB9603095D0 (en) 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline derivatives
EP0885198B1 (fr) 1996-03-05 2001-12-19 AstraZeneca AB Derives de 4-anilinoquinazoline
EP0912559B1 (fr) 1996-07-13 2002-11-06 Glaxo Group Limited Composes heterocycliques condenses en tant qu'inhibiteurs de la proteine tyrosine kinase
CN1230185A (zh) 1996-07-13 1999-09-29 葛兰素集团有限公司 双环芳杂环化合物用作蛋白质酪氨酸激酶的抑制剂
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
ES2175415T3 (es) 1996-07-18 2002-11-16 Pfizer Inhibidores de metaloproteasas matriciales basados en fosfinato.
EA199900139A1 (ru) 1996-08-23 1999-08-26 Пфайзер, Инк. Производные арилсульфониламиногидроксамовой кислоты
PT950059E (pt) 1997-01-06 2004-10-29 Pfizer Derivados de sulfona ciclicos
EA002594B1 (ru) 1997-02-03 2002-06-27 Пфайзер Продактс Инк. Производные арилсульфониламиногидроксамовой кислоты
CA2279863A1 (fr) 1997-02-07 1998-08-13 Pfizer Inc. Derives du n-hxdroxy-beta-sulfonyl-propionamide et leur utilisation comme inhibiteurs des metalloproteases matrices
JP3710489B2 (ja) 1997-02-11 2005-10-26 ファイザー・インク アリールスルホニルヒドロキサム酸誘導体
JP2002511852A (ja) 1997-05-07 2002-04-16 スージェン・インコーポレーテッド 蛋白質キナーゼ活性の調節剤としての2−インドリノン誘導体
AU734009B2 (en) 1997-05-30 2001-05-31 Merck & Co., Inc. Novel angiogenesis inhibitors
ES2289791T3 (es) 1997-08-22 2008-02-01 Astrazeneca Ab Derivados de oxindolilquinazolina como inhibidores de la angiogenesis.
WO1999016755A1 (fr) 1997-09-26 1999-04-08 Merck & Co., Inc. Nouveaux inhibiteurs de l'angiogenese
WO1999024440A1 (fr) 1997-11-11 1999-05-20 Pfizer Products Inc. Derives de thienopyrimidine et thienopyridine utiles comme agents anticancereux
GB9725782D0 (en) 1997-12-05 1998-02-04 Pfizer Ltd Therapeutic agents
RS49779B (sr) 1998-01-12 2008-06-05 Glaxo Group Limited, Biciklična heteroaromatična jedinjenja kao inhibitori protein tirozin kinaze
GB9800575D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
GB9801690D0 (en) 1998-01-27 1998-03-25 Pfizer Ltd Therapeutic agents
PA8469401A1 (es) 1998-04-10 2000-05-24 Pfizer Prod Inc Derivados biciclicos del acido hidroxamico
PA8469501A1 (es) 1998-04-10 2000-09-29 Pfizer Prod Inc Hidroxamidas del acido (4-arilsulfonilamino)-tetrahidropiran-4-carboxilico
AU759226B2 (en) 1998-05-29 2003-04-10 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
UA71945C2 (en) 1999-01-27 2005-01-17 Pfizer Prod Inc Substituted bicyclic derivatives being used as anticancer agents
JP3270834B2 (ja) 1999-01-27 2002-04-02 ファイザー・プロダクツ・インク 抗がん剤として有用なヘテロ芳香族二環式誘導体
DK1292591T3 (da) 2000-06-22 2005-05-30 Pfizer Prod Inc Bicykliske derivater til behandling af abnorm cellevækst
AR042586A1 (es) 2001-02-15 2005-06-29 Sugen Inc 3-(4-amidopirrol-2-ilmetiliden)-2-indolinona como inhibidores de la protein quinasa; sus composiciones farmaceuticas; un metodo para la modulacion de la actividad catalitica de la proteinquinasa; un metodo para tratar o prevenir una afeccion relacionada con la proteinquinasa
JP2008536950A (ja) * 2005-04-18 2008-09-11 ニューロジェン・コーポレーション 置換ヘテロアリールのcb1拮抗薬
US20090281089A1 (en) * 2008-04-11 2009-11-12 Genentech, Inc. Pyridyl inhibitors of hedgehog signalling
MX2010012457A (es) * 2008-05-13 2010-12-07 Irm Llc Heterociclos que contienen nitrogeno fusionado y composiciones de los mismos como inhibidores de cinasa.
GB0813740D0 (en) * 2008-07-28 2008-09-03 Angeletti P Ist Richerche Biologica Therapeutic compounds
US9665874B2 (en) 2012-03-13 2017-05-30 American Express Travel Related Services Company, Inc. Systems and methods for tailoring marketing
US10884952B2 (en) 2016-09-30 2021-01-05 Intel Corporation Enforcing memory operand types using protection keys
JP6943759B2 (ja) 2017-12-28 2021-10-06 株式会社東海理化電機製作所 シフト装置

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100029615A1 (en) 2006-12-15 2010-02-04 Michael John Munchhof Benzimidazole derivatives
US8148401B2 (en) 2007-06-29 2012-04-03 Pfizer Inc. Benzimidazole derivatives
US8431597B2 (en) 2007-06-29 2013-04-30 Pfizer Inc. Benzimidazole derivatives
WO2009154739A2 (fr) 2008-06-17 2009-12-23 Duke University Modulateurs du récepteur smoothened
US20120157471A1 (en) 2009-09-01 2012-06-21 Pfizer Inc. Benzimidazole derivatives

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
Alcedo et al., "The Drosophila smoothened gene encodes a seven-pass membrane protein, a putative receptor for the hedgehog signal," Cell 86: 221-232 (1996).
Chen and Struhl, "Dual roles for patched in sequestering and transducing Hedgehog," Cell 87: 553-63 (1996).
Chidambaram et al., "Mutations in the human homologue of the Drosophila patched gene in Caucasian and African-American nevoid basal cell carcinoma syndrome patients," Cancer Research 56: 4599-601 (1996).
Gailani et al., "The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas," Nature Genet. 14: 78-81 (1996).
Hahn et al., "Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome," Cell 85: 841-51 (1996).
Hooper and Scott, "The Drosophila patched gene encodes a putative membrane protein required for segmental patterning," Cell 59: 751-65 (1989).
International Preliminary Report on Patentability for PCT/IB2011/054675, issued Apr. 23, 2013.
International Search Report for PCT/IB2011/054675, mailed Dec. 14, 2011.
Johnson et al., "Human homolog of patched, a candidate gene for the basal cell nevus syndrome," Science 272: 1668-71 (1996).
Marigo et al., "Biochemical evidence that patched is the Hedgehog receptor," Nature 384: 176-9 (1996).
Nakano et al., "A protein with several possible membrane-spanning domains encoded by the Drosophila segment polarity gene patched," Nature 341: 508-13 (1989).
Rohner et al., "Effective Targeting of Hedgehog Signaling in a Medulloblastoma Model with PF-5274857, a Potent and Selective Smoothened Antagonist That Penetrates the Blood-Brain Barrier," Mol. Cancer Ther., 2012; 11:57-65, published online Nov. 14, 2011.
Stevens G et al., "Toxicologic Characterization of PF-05274857, a Potent and Selective Smoothened Inhibitor for the Treatment of Advanced Cancer," Abstract No. 2268, Poster No. 226, presented at Society of Toxicology Annual Meeting 2012 on Mar 14, 2012.
Stone et al., "The tumour-suppressor gene patched encodes a candidate receptor for Sonic hedgehog," Nature 384:129-34 (1996).
Unden et al., "Mutations in the human homologue of Drosophila patched (PTCH) in basal cell carcinomas and the Gorlin syndrome: different in vivo mechanisms of PTCH inactivation," Cancer Res. 56: 4562-4565 (1996).
van den Heuvel & Ingham, "Smoothened encodes a receptor-like serpentine protein required for hedgehog signaling," Nature 382: 547-551 (1996).
Wicking et al., "Most germ-line mutations in the nevoid basal cell carcinoma syndrome lead to a premature termination of the Patched protein, and no genotype-phenotype correlations are evident," Am. J. Hum. Genet. 60: 21-6 (1997).
Xie et al., "Activating Smoothened mutations in sporadic basal-cell carcinoma," Nature 391: 90-92 (1998).

Also Published As

Publication number Publication date
ES2543151T3 (es) 2015-08-17
JP5802756B2 (ja) 2015-11-04
EP2630134B1 (fr) 2015-05-20
CA2813162C (fr) 2015-06-16
EP2630134A1 (fr) 2013-08-28
EP2630134B9 (fr) 2018-04-18
US20130210800A1 (en) 2013-08-15
CA2813162A1 (fr) 2012-04-26
WO2012052948A1 (fr) 2012-04-26
JP2013543842A (ja) 2013-12-09
ES2543151T9 (es) 2018-08-24

Similar Documents

Publication Publication Date Title
US9056865B2 (en) Pyridine-2-derivatives as smoothened receptor modulators
US20120157471A1 (en) Benzimidazole derivatives
US8431597B2 (en) Benzimidazole derivatives
US9290496B2 (en) Purine derivatives
US20100029615A1 (en) Benzimidazole derivatives
US10676471B2 (en) Cycloalkyl-linked diheterocycle derivatives
US9758538B2 (en) Pyrimidine derivatives
WO2015155624A1 (fr) Dérivés de dihydropyrrolopyrimidine
WO2016001789A1 (fr) Dérivés de pyrimidine en tant qu'inhibiteurs de pi3k destinés à être utilisés dans le traitement du cancer
OA18105A (en) Cycloalkyl-linked diheterocycle derivatives

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20230616